1
|
Dufford MT, Fleischer TC, Sommerville LJ, Badsha MB, Polpitiya AD, Logan J, Fox AC, Rust SR, Cox CB, Garite TJ, Boniface JJ, Kearney PE. Clock Proteins Have the Potential to Improve Term Delivery Date Prediction: A Proof-of-Concept Study. Life (Basel) 2025; 15:224. [PMID: 40003633 PMCID: PMC11856609 DOI: 10.3390/life15020224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Our ability to accurately predict the delivery date of term pregnancies is limited by shortcomings of modern-day clinical tools and due date estimation methods. The pregnancy clock is a series of coordinated and harmonized signals between mother, fetus, and placenta that regulate the length of gestation. Clock proteins are thought to be important mediators of these signals, yet few studies have investigated their potential utility as predictors of term delivery date. In this study, we performed a cross-sectional proteome analysis of 2648 serum samples collected between 18 and 28 weeks of gestation from mothers who delivered at term. The cohort included pregnancies both with and without complications. A total of 15 proteins of diverse functionalities were shown to have a direct association with time to birth (TTB), 11 of which have not been previously linked to gestational age. The protein A Distintegrin and Metalloproteinase 12 (ADA12) was one of the 15 proteins shown to have an association with TTB. Mothers who expressed the highest levels of ADA12 in the cohort (90th percentile) gave birth earlier than mothers who expressed the lowest levels of ADA12 (10th percentile) at a statistically significant rate (median gestational age at birth 390/7 weeks vs. 393/7 weeks, p < 0.001). Altogether, these findings suggest that ADA12, as well as potentially other clock proteins, have the potential to serve as clinical predictors of term delivery date in uncomplicated pregnancies and represent an important step towards characterizing the role(s) of clock proteins in mediating pregnancy length.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Paul E. Kearney
- Sera Prognostics, Inc., Salt Lake City, UT 84109, USA (T.C.F.); (A.C.F.); (T.J.G.)
| |
Collapse
|
2
|
Zeng X, Cai Y, Wu M, Chen H, Sun M, Yang H. An overview of current advances in perinatal alcohol exposure and pathogenesis of fetal alcohol spectrum disorders. J Neurodev Disord 2024; 16:20. [PMID: 38643092 PMCID: PMC11031898 DOI: 10.1186/s11689-024-09537-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/08/2024] [Indexed: 04/22/2024] Open
Abstract
The adverse use of alcohol is a serious global public health problem. Maternal alcohol consumption during pregnancy usually causes prenatal alcohol exposure (PAE) in the developing fetus, leading to a spectrum of disorders known as fetal alcohol spectrum disorders (FASD) and even fetal alcohol syndrome (FAS) throughout the lifelong sufferers. The prevalence of FASD is approximately 7.7 per 1,000 worldwide, and is even higher in developed regions. Generally, Ethanol in alcoholic beverages can impair embryonic neurological development through multiple pathways leading to FASD. Among them, the leading mechanism of FASDs is attributed to ethanol-induced neuroinflammatory damage to the central nervous system (CNS). Although the underlying molecular mechanisms remain unclear, the remaining multiple pathological mechanisms is likely due to the neurotoxic damage of ethanol and the resultant neuronal loss. Regardless of the molecular pathway, the ultimate outcome of the developing CNS exposed to ethanol is almost always the destruction and apoptosis of neurons, which leads to the reduction of neurons and further the development of FASD. In this review, we systematically summarize the current research progress on the pathogenesis of FASD, which hopefully provides new insights into differential early diagnosis, treatment and prevention for patents with FASD.
Collapse
Affiliation(s)
- Xingdong Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Yongle Cai
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Mengyan Wu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Haonan Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China.
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
3
|
Andrade-Brito DE, Núñez-Ríos DL, Martínez-Magaña JJ, Nagamatsu ST, Rompala G, Zillich L, Witt SH, Clark SL, Lattig MC, Montalvo-Ortiz JL. Neuronal-specific methylome and hydroxymethylome analysis reveal significant loci associated with alcohol use disorder. Front Genet 2024; 15:1345410. [PMID: 38633406 PMCID: PMC11021708 DOI: 10.3389/fgene.2024.1345410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/14/2024] [Indexed: 04/19/2024] Open
Abstract
Background: Alcohol use disorder (AUD) is a complex condition associated with adverse health consequences that affect millions of individuals worldwide. Epigenetic modifications, including DNA methylation (5 mC), have been associated with AUD and other alcohol-related traits. Epigenome-wide association studies (EWAS) have identified differentially methylated genes associated with AUD in human peripheral and brain tissue. More recently, epigenetic studies of AUD have also evaluated DNA hydroxymethylation (5 hmC) in the human brain. However, most of the epigenetic work in postmortem brain tissue has examined bulk tissue. In this study, we investigated neuronal-specific 5 mC and 5 hmC alterations at CpG sites associated with AUD in the human orbitofrontal cortex (OFC). Methods: Neuronal nuclei from the OFC were evaluated in 34 human postmortem brain samples (10 AUD, 24 non-AUD). Reduced representation oxidative bisulfite sequencing was used to assess 5 mC and 5 hmC at the genome-wide level. Differential 5 mC and 5 hmC were evaluated using the methylKit R package and significance was set at false discovery rate < 0.05 and differential methylation > 2. Functional enrichment analyses were performed, and gene-level convergence was evaluated in an independent dataset that assessed 5 mC and 5 hmC of AUD in bulk cortical tissue. Results: We identified 417 5 mC and 363 5hmC significant differential CpG sites associated with AUD, with 59% in gene promoters. Some of the identified genes have been previously implicated in alcohol consumption, including SYK, DNMT3A for 5 mC, GAD1, DLX1, DLX2, for 5 hmC and GATA4 in both. Convergence with a previous AUD 5 mC and 5 hmC study was observed for 28 genes. We also identified 5 and 35 differential regions for 5 mC and 5 hmC, respectively. Lastly, GWAS enrichment analysis showed an association with AUD for differential 5 mC genes. Discussion: This study reveals neuronal-specific methylome and hydroxymethylome dysregulation associated with AUD, identifying both previously reported and potentially novel gene associations with AUD. Our findings provide new insights into the epigenomic dysregulation of AUD in the human brain.
Collapse
Affiliation(s)
- Diego E. Andrade-Brito
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| | - Diana L. Núñez-Ríos
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| | - José Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| | - Sheila T. Nagamatsu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| | - Gregory Rompala
- Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H. Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Shaunna L. Clark
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, College Station, TX, United States
| | - Maria C. Lattig
- Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Janitza L. Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| |
Collapse
|
4
|
Andrade-Brito DE, Núñez-Ríos DL, Martínez-Magaña JJ, Nagamatsu ST, Rompala G, Zillich L, Witt SH, Clark SL, Latig MC, Traumatic Stress Brain Research Group, PGC SUD Epigenetics Working Group, Montalvo-Ortiz JL. Neuronal-specific methylome and hydroxymethylome analysis reveal replicated and novel loci associated with alcohol use disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.28.23299094. [PMID: 38105948 PMCID: PMC10725575 DOI: 10.1101/2023.11.28.23299094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Alcohol use disorder (AUD) is a complex condition associated with adverse health consequences that affect millions of individuals worldwide. Epigenetic modifications, including DNA methylation (5mC), have been associated with AUD and other alcohol-related traits. Epigenome-wide association studies (EWAS) have identified differentially methylated genes associated with AUD in human peripheral and brain tissue. More recently, epigenetic studies of AUD have also evaluated DNA hydroxymethylation (5hmC) in the human brain. However, most of the epigenetic work in postmortem brain tissue has examined bulk tissue. In this study, we investigated neuronal-specific 5mC and 5hmC alterations at CpG sites associated with AUD in the human orbitofrontal cortex (OFC). Neuronal nuclei from the OFC were evaluated in 34 human postmortem brain samples (10 AUD, 24 non-AUD). Reduced representation oxidative bisulfite sequencing was used to assess 5mC and 5hmC at the genome-wide level. Differential 5mC and 5hmC were evaluated using the methylKit R package and significance was set at false discovery rate <0.05 and differential methylation >2. Functional enrichment analyses were performed and replication was evaluated replication in an independent dataset that assessed 5mC and 5hmC of AUD in bulk cortical tissue. We identified 417 5mC and 363 5hmC genome-wide significant differential CpG sites associated with AUD, with 59% in gene promoters. We also identified genes previously implicated in alcohol consumption, such as SYK, CHRM2, DNMT3A, and GATA4, for 5mC and GATA4, and GAD1, GATA4, DLX1 for 5hmC. Replication was observed for 28 CpG sites from a previous AUD 5mC and 5hmC study, including FOXP1. Lastly, GWAS enrichment analysis showed an association with AUD for differential 5mC genes. This study reveals neuronal-specific methylome and hydroxymethylome dysregulation associated with AUD. We replicated previous findings and identified novel associations with AUD for both 5mC and 5hmC marks within the OFC. Our findings provide new insights into the epigenomic dysregulation of AUD in the human brain.
Collapse
Affiliation(s)
- Diego E. Andrade-Brito
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| | - Diana L. Núñez-Ríos
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| | - José Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| | - Sheila T. Nagamatsu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| | - Gregory Rompala
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H. Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Shaunna L. Clark
- Department of Psychiatry & Behavioral Sciences, Texas A&M University, College Station, Texas, USA
| | - Maria C. Latig
- Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | | | - Janitza L. Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| |
Collapse
|
5
|
Popova S, Charness ME, Burd L, Crawford A, Hoyme HE, Mukherjee RAS, Riley EP, Elliott EJ. Fetal alcohol spectrum disorders. Nat Rev Dis Primers 2023; 9:11. [PMID: 36823161 DOI: 10.1038/s41572-023-00420-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 02/25/2023]
Abstract
Alcohol readily crosses the placenta and may disrupt fetal development. Harm from prenatal alcohol exposure (PAE) is determined by the dose, pattern, timing and duration of exposure, fetal and maternal genetics, maternal nutrition, concurrent substance use, and epigenetic responses. A safe dose of alcohol use during pregnancy has not been established. PAE can cause fetal alcohol spectrum disorders (FASD), which are characterized by neurodevelopmental impairment with or without facial dysmorphology, congenital anomalies and poor growth. FASD are a leading preventable cause of birth defects and developmental disability. The prevalence of FASD in 76 countries is >1% and is high in individuals living in out-of-home care or engaged in justice and mental health systems. The social and economic effects of FASD are profound, but the diagnosis is often missed or delayed and receives little public recognition. Future research should be informed by people living with FASD and be guided by cultural context, seek consensus on diagnostic criteria and evidence-based treatments, and describe the pathophysiology and lifelong effects of FASD. Imperatives include reducing stigma, equitable access to services, improved quality of life for people with FASD and FASD prevention in future generations.
Collapse
Affiliation(s)
- Svetlana Popova
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada.
| | - Michael E Charness
- VA Boston Healthcare System, West Roxbury, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA.,Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.,Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Larry Burd
- North Dakota Fetal Alcohol Syndrome Center, Department of Pediatrics, University of North Dakota School of Medicine and Health Sciences, Pediatric Therapy Services, Altru Health System, Grand Forks, ND, USA
| | - Andi Crawford
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - H Eugene Hoyme
- Sanford Children's Genomic Medicine Consortium, Sanford Health, and University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA
| | - Raja A S Mukherjee
- National UK FASD Clinic, Surrey and Borders Partnership NHS Foundation Trust, Redhill, Surrey, UK
| | - Edward P Riley
- Center for Behavioral Teratology, San Diego State University, San Diego, CA, USA
| | - Elizabeth J Elliott
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,New South Wales FASD Assessment Service, CICADA Centre for Care and Intervention for Children and Adolescents affected by Drugs and Alcohol, Sydney Children's Hospitals Network, Westmead, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Blood Vessels as a Key Mediator for Ethanol Toxicity: Implication for Neuronal Damage. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111882. [PMID: 36431016 PMCID: PMC9696276 DOI: 10.3390/life12111882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Excessive intake of ethanol is associated with severe brain dysfunction, and the subsequent neurological and behavioral abnormalities are well-established social risks. Many research studies have addressed how ethanol induces neurological toxicity. However, the underlying mechanisms with which ethanol induces neurological toxicity are still obscure, perhaps due to the variety and complexity of these mechanisms. Epithelial cells are in direct contact with blood and can thus mediate ethanol neurotoxicity. Ethanol activates the endothelial cells of blood vessels, as well as lymphatic vessels, in a concentration-dependent manner. Among various signaling mediators, nitric oxide plays important roles in response to ethanol. Endothelial and inducible nitric oxide synthases (eNOS and iNOS) are upregulated and activated by ethanol and enhance neuroinflammation. On the other hand, angiogenesis and blood vessel remodeling are both affected by ethanol intake, altering blood supply and releasing angiocrine factors to regulate neuronal functions. Thus, ethanol directly acts on endothelial cells, yet the molecular target(s) on endothelial cells remain unknown. Previous studies on neurons and glial cells have validated the potential contribution of membrane lipids and some specific proteins as ethanol targets, which may also be the case in endothelial cells. Future studies, based on current knowledge, will allow for a greater understanding of the contribution and underlying mechanisms of endothelial cells in ethanol-induced neurological toxicity, protecting neurological health against ethanol toxicity.
Collapse
|
7
|
TRPV6 channel mediates alcohol-induced gut barrier dysfunction and systemic response. Cell Rep 2022; 39:110937. [PMID: 35705057 PMCID: PMC9250449 DOI: 10.1016/j.celrep.2022.110937] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/20/2022] [Accepted: 05/18/2022] [Indexed: 11/22/2022] Open
Abstract
Intestinal epithelial tight junction disruption is a primary contributing factor in alcohol-associated endotoxemia, systemic inflammation, and multiple organ damage. Ethanol and acetaldehyde disrupt tight junctions by elevating intracellular Ca2+. Here we identify TRPV6, a Ca2+-permeable channel, as responsible for alcohol-induced elevation of intracellular Ca2+, intestinal barrier dysfunction, and systemic inflammation. Ethanol and acetaldehyde elicit TRPV6 ionic currents in Caco-2 cells. Studies in Caco-2 cell monolayers and mouse intestinal organoids show that TRPV6 deficiency or inhibition attenuates ethanol- and acetaldehyde-induced Ca2+ influx, tight junction disruption, and barrier dysfunction. Moreover, Trpv6−/− mice are resistant to alcohol-induced intestinal barrier dysfunction. Photoaffinity labeling of 3-azibutanol identifies a histidine as a potential alcohol-binding site in TRPV6. The substitution of this histidine, and a nearby arginine, reduces ethanol-activated currents. Our findings reveal that TRPV6 is required for alcohol-induced gut barrier dysfunction and inflammation. Molecules that decrease TRPV6 function have the potential to attenuate alcohol-associated tissue injury. Meena et al. show that the mechanism of alcohol-induced gut permeability, endotoxemia, and systemic inflammation requires the TRPV6 channel. They show that ethanol activates TRPV6, induces calcium influx, and disrupts intestinal epithelial tight junctions. Furthermore, specific histidine and arginine residues at the N terminus fine-tune the alcohol-induced activation of TRPV6.
Collapse
|
8
|
Abstract
This article is part of a Festschrift commemorating the 50th anniversary of the National Institute on Alcohol Abuse and Alcoholism (NIAAA). Established in 1970, first as part of the National Institute of Mental Health and later as an independent institute of the National Institutes of Health, NIAAA today is the world’s largest funding agency for alcohol research. In addition to its own intramural research program, NIAAA supports the entire spectrum of innovative basic, translational, and clinical research to advance the diagnosis, prevention, and treatment of alcohol use disorder and alcohol-related problems. To celebrate the anniversary, NIAAA hosted a 2-day symposium, “Alcohol Across the Lifespan: 50 Years of Evidence-Based Diagnosis, Prevention, and Treatment Research,” devoted to key topics within the field of alcohol research. This article is based on Dr. Charness’ presentation at the event. NIAAA Director George F. Koob, Ph.D., serves as editor of the Festschrift.
Collapse
Affiliation(s)
- Michael E Charness
- VA Boston Healthcare System, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
9
|
Licheri V, Brigman JL. Altering Cell-Cell Interaction in Prenatal Alcohol Exposure Models: Insight on Cell-Adhesion Molecules During Brain Development. Front Mol Neurosci 2022; 14:753537. [PMID: 34975396 PMCID: PMC8715949 DOI: 10.3389/fnmol.2021.753537] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Alcohol exposure during pregnancy disrupts the development of the brain and produces long lasting behavioral and cognitive impairments collectively known as Fetal Alcohol Spectrum Disorders (FASDs). FASDs are characterized by alterations in learning, working memory, social behavior and executive function. A large body of literature using preclinical prenatal alcohol exposure models reports alcohol-induced changes in architecture and activity in specific brain regions affecting cognition. While multiple putative mechanisms of alcohol’s long-lasting effects on morphology and behavior have been investigated, an area that has received less attention is the effect of alcohol on cell adhesion molecules (CAMs). The embryo/fetal development represents a crucial period for Central Nervous System (CNS) development during which the cell-cell interaction plays an important role. CAMs play a critical role in neuronal migration and differentiation, synaptic organization and function which may be disrupted by alcohol. In this review, we summarize the physiological structure and role of CAMs involved in brain development, review the current literature on prenatal alcohol exposure effects on CAM function in different experimental models and pinpoint areas needed for future study to better understand how CAMs may mediate the morphological, sensory and behavioral outcomes in FASDs.
Collapse
Affiliation(s)
- Valentina Licheri
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States.,New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
10
|
Drake DM, Wells PG. Novel mechanisms in alcohol neurodevelopmental disorders via BRCA1 depletion and BRCA1-dependent NADPH oxidase regulation. Redox Biol 2021; 48:102148. [PMID: 34736119 PMCID: PMC8577473 DOI: 10.1016/j.redox.2021.102148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 11/27/2022] Open
Abstract
The breast cancer 1 protein (BRCA1) facilitates DNA repair, preventing embryolethality and protecting the fetus from reactive oxygen species (ROS)-induced developmental disorders mediated by oxidatively damaged DNA. Alcohol (ethanol, EtOH) exposure during pregnancy causes fetal alcohol spectrum disorders (FASD), characterized by aberrant behaviour and enhanced ROS formation and proteasomal protein degradation. Herein, ROS-producing NADPH oxidase (NOX) activity was higher in Brca1 +/- vs. +/+ fetal and adult brains, and further enhanced by a single EtOH exposure. EtOH also enhanced catalase and proteasomal activities, while conversely reducing BRCA1 protein levels without affecting Brca1 gene expression. EtOH-initiated adaptive postnatal freezing behaviour was lost in Brca1 +/- progeny. Pretreatment with the free radical spin trap and ROS inhibitor phenylbutylnitrone blocked all EtOH effects, suggesting ROS-dependent mechanisms. This is the first in vivo evidence of NOX regulation by BRCA1, and of EtOH-induced, ROS-mediated depletion of BRCA1, revealing novel mechanisms of BRCA1 protection in FASD.
Collapse
Affiliation(s)
- Danielle M Drake
- Department of Pharmaceutical Sciences and Centre for Pharmaceutical Oncology, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Peter G Wells
- Department of Pharmaceutical Sciences and Centre for Pharmaceutical Oncology, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Boschen KE, Ptacek TS, Simon JM, Parnell SE. Transcriptome-Wide Regulation of Key Developmental Pathways in the Mouse Neural Tube by Prenatal Alcohol Exposure. Alcohol Clin Exp Res 2020; 44:1540-1550. [PMID: 32557641 DOI: 10.1111/acer.14389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/02/2020] [Accepted: 05/31/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Early gestational alcohol exposure is associated with severe craniofacial and CNS dysmorphologies and behavioral abnormalities during adolescence and adulthood. Alcohol exposure during the formation of the neural tube (gestational day [GD] 8 to 10 in mice; equivalent to4th week of human pregnancy) disrupts development of ventral midline brain structures such as the pituitary, septum, and ventricles. This study identifies transcriptomic changes in the rostroventral neural tube (RVNT), the region of the neural tube that gives rise to the midline structures sensitive to alcohol exposure during neurulation. METHODS Female C57BL/6J mice were administered 2 doses of alcohol (2.9 g/kg) or vehicle 4 hours apart on GD 9.0. The RVNTs of embryos were collected 6 or 24 hours after the first dose and processed for RNA-seq. RESULTS Six hours following GD 9.0 alcohol exposure (GD 9.25), over 2,300 genes in the RVNT were determined to be differentially regulated by alcohol. Enrichment analysis determined that PAE affected pathways related to cell proliferation, p53 signaling, ribosome biogenesis, and immune activation. In addition, over 100 genes involved in primary cilia formation and function and regulation of morphogenic pathways were altered 6 hours after alcohol exposure. The changes to gene expression were largely transient, as only 91 genes identified as differentially regulated by prenatal alcohol at GD 10 (24 hours postexposure). Functionally, the differentially regulated genes at GD 10 were related to organogenesis and cell migration. CONCLUSIONS These data give a comprehensive view of the changing landscape of the embryonic transcriptome networks in regions of the neural tube that give rise to brain structures impacted by a neurulation-stage alcohol exposure. Identification of gene networks dysregulated by alcohol will help elucidate the pathogenic mechanisms of alcohol's actions.
Collapse
Affiliation(s)
- Karen E Boschen
- From the Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Travis S Ptacek
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jeremy M Simon
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Scott E Parnell
- From the Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
12
|
You Y, Das J. Effect of ethanol on Munc13-1 C1 in Membrane: A Molecular Dynamics Simulation Study. Alcohol Clin Exp Res 2020; 44:1344-1355. [PMID: 32424866 DOI: 10.1111/acer.14363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 05/06/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND EtOH has a significant effect on synaptic plasticity. Munc13-1 is an essential presynaptic active zone protein involved in priming the synaptic vesicle and releasing neurotransmitter in the brain. It is a peripheral membrane protein and binds to the activator, diacylglycerol (DAG)/phorbol ester at its membrane-targeting C1 domain. Our previous studies identified Glu-582 of C1 domain as the alcohol-binding residue (Das, J. et al, J. Neurochem., 126, 715-726, 2013). METHODS Here, we describe a 250 ns molecular dynamics (MD) simulation study on the interaction of EtOH and the activator-bound Munc13-1 C1 in the presence of varying concentrations of phosphatidylserine (PS). RESULTS In this study, Munc13-1 C1 shows higher conformational stability in EtOH than in water. It forms fewer hydrogen bonds with phorbol 13-acetate in the presence of EtOH than in water. EtOH also affected the interaction between the protein and the membrane and between the activator and the membrane. Similar studies in a E582A mutant suggest that these effects of EtOH are mostly mediated through Glu-582. CONCLUSIONS EtOH forms hydrogen bonds with Glu-582. While occupancy of the EtOH molecules at the vicinity (4Å) of Glu-582 is 34.4%, the occupancy in the E582A mutant is 26.5% of the simulation time. In addition, the amount of PS in the membrane influences the conformational stability of the C1 domain and interactions in the ternary complex. This study is important in providing the structural basis of EtOH's effects on synaptic plasticity.
Collapse
Affiliation(s)
- Youngki You
- From the Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Joydip Das
- From the Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
13
|
Dou X, Lee JY, Charness ME. Neuroprotective Peptide NAPVSIPQ Antagonizes Ethanol Inhibition of L1 Adhesion by Promoting the Dissociation of L1 and Ankyrin-G. Biol Psychiatry 2020; 87:656-665. [PMID: 31640849 PMCID: PMC7056560 DOI: 10.1016/j.biopsych.2019.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Ethanol causes developmental neurotoxicity partly by blocking adhesion mediated by the L1 neural cell adhesion molecule. This action of ethanol is antagonized by femtomolar concentrations of the neuropeptide NAPVSIPQ (NAP), an active fragment of the activity-dependent neuroprotective protein (ADNP). How femtomolar concentrations of NAP antagonize millimolar concentrations of ethanol is unknown. L1 sensitivity to ethanol requires L1 association with ankyrin-G; therefore, we asked whether NAP promotes the dissociation of ankyrin-G and L1. METHODS L1-ankyrin-G association was studied using immunoprecipitation, Western blotting, and immunofluorescence in NIH/3T3 cells transfected with wild-type and mutated human L1 genes. Phosphorylation of the ankyrin binding motif in the L1 cytoplasmic domain was studied after NAP treatment of intact cells, rat brain homogenates, and purified protein fragments. RESULTS Femtomolar concentrations of NAP stimulated the phosphorylation of tyrosine-1229 (L1-Y1229) at the ankyrin binding motif of the L1 cytoplasmic domain, leading to the dissociation of L1 from ankyrin-G and the spectrin-actin cytoskeleton. NAP increased the association of L1 and EphB2 and directly activated EphB2 phosphorylation of L1-Y1229. These actions of NAP were reproduced by P7A-NAP, a NAP variant that also blocks the teratogenic actions of ethanol, but not by I6A-NAP, which does not block ethanol teratogenesis as potently. Finally, knockdown of EPHB2 prevented ethanol inhibition of L1 adhesion in NIH/3T3 cells. CONCLUSIONS NAP potently antagonizes ethanol inhibition of L1 adhesion by stimulating EphB2 phosphorylation of L1-Y1229. EphB2 plays a critical role in synaptic development; its potent activation by NAP suggests that ADNP may mediate synaptic development partly by activating EphB2.
Collapse
Affiliation(s)
- Xiaowei Dou
- Veterans Affairs Boston Healthcare System; Department of Neurology, Harvard Medical School, West Roxbury, MA 02132
| | - Jerry Y. Lee
- Veterans Affairs Boston Healthcare System; Department of Neurology, Harvard Medical School, West Roxbury, MA 02132
| | - Michael E. Charness
- Veterans Affairs Boston Healthcare System; Department of Neurology, Harvard Medical School, West Roxbury, MA 02132,Department of Neurology, Boston University, School of Medicine, Boston, MA 02119, To whom correspondence should be addressed. Michael E. Charness, M.D., VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, Phone: 857-203-6011,
| |
Collapse
|
14
|
Das J. SNARE Complex-Associated Proteins and Alcohol. Alcohol Clin Exp Res 2019; 44:7-18. [PMID: 31724225 DOI: 10.1111/acer.14238] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/07/2019] [Indexed: 12/23/2022]
Abstract
Alcohol addiction causes major health problems throughout the world, causing numerous deaths and incurring a huge economic burden to society. To develop an intervention for alcohol addiction, it is necessary to identify molecular target(s) of alcohol and associated molecular mechanisms of alcohol action. The functions of many central and peripheral synapses are impacted by low concentrations of ethanol (EtOH). While the postsynaptic targets and mechanisms are studied extensively, there are limited studies on the presynaptic targets and mechanisms. This article is an endeavor in this direction, focusing on the effect of EtOH on the presynaptic proteins associated with the neurotransmitter release machinery. Studies on the effects of EtOH at the levels of gene, protein, and behavior are highlighted in this article.
Collapse
Affiliation(s)
- Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
15
|
Bhatia S, Drake DM, Miller L, Wells PG. Oxidative stress and DNA damage in the mechanism of fetal alcohol spectrum disorders. Birth Defects Res 2019; 111:714-748. [PMID: 31033255 DOI: 10.1002/bdr2.1509] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/18/2022]
Abstract
This review covers molecular mechanisms involving oxidative stress and DNA damage that may contribute to morphological and functional developmental disorders in animal models resulting from exposure to alcohol (ethanol, EtOH) in utero or in embryo culture. Components covered include: (a) a brief overview of EtOH metabolism and embryopathic mechanisms other than oxidative stress; (b) mechanisms within the embryo and fetal brain by which EtOH increases the formation of reactive oxygen species (ROS); (c) critical embryonic/fetal antioxidative enzymes and substrates that detoxify ROS; (d) mechanisms by which ROS can alter development, including ROS-mediated signal transduction and oxidative DNA damage, the latter of which leads to pathogenic genetic (mutations) and epigenetic changes; (e) pathways of DNA repair that mitigate the pathogenic effects of DNA damage; (f) related indirect mechanisms by which EtOH enhances risk, for example by enhancing the degradation of some DNA repair proteins; and, (g) embryonic/fetal pathways like NRF2 that regulate the levels of many of the above components. Particular attention is paid to studies in which chemical and/or genetic manipulation of the above mechanisms has been shown to alter the ability of EtOH to adversely affect development. Alterations in the above components are also discussed in terms of: (a) individual embryonic and fetal determinants of risk and (b) potential risk biomarkers and mitigating strategies. FASD risk is likely increased in progeny which/who are biochemically predisposed via genetic and/or environmental mechanisms, including enhanced pathways for ROS formation and/or deficient pathways for ROS detoxification or DNA repair.
Collapse
Affiliation(s)
- Shama Bhatia
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Danielle M Drake
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada
| | | | - Peter G Wells
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, University of Toronto, Toronto, Ontario, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Das J. Identification of alcohol-binding site(s) in proteins using diazirine-based photoaffinity labeling and mass spectrometry. Chem Biol Drug Des 2018; 93:1158-1165. [PMID: 30346111 DOI: 10.1111/cbdd.13403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/03/2018] [Accepted: 09/15/2018] [Indexed: 01/12/2023]
Abstract
Defining molecular targets of alcohol and understanding the molecular mechanism of alcohol actions are necessary to develop effective therapeutics for alcohol use disorder (AUD). Here, we describe a detailed protocol for identifying alcohol-binding site(s) in proteins using diazirine-based azialcohol as photoaffinity labeling agents. Upon photoirradiation, azialcohol photoincorporates into alcohol-binding proteins. The stoichiometry and site of azialcohol photoincorporation can be determined using high-resolution mass spectrometry. Identification of the alcohol-binding residues in protein followed by measuring the biological significance of these residues in regulating alcohol action are important steps in characterizing the molecular targets of alcohol.
Collapse
Affiliation(s)
- Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
17
|
Dou X, Menkari C, Mitsuyama R, Foroud T, Wetherill L, Hammond P, Suttie M, Chen X, Chen SY, Charness ME. L1 coupling to ankyrin and the spectrin-actin cytoskeleton modulates ethanol inhibition of L1 adhesion and ethanol teratogenesis. FASEB J 2018; 32:1364-1374. [PMID: 29109170 DOI: 10.1096/fj.201700970] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Ethanol causes fetal alcohol spectrum disorders (FASDs) partly by inhibiting cell adhesion mediated by the L1 neural cell adhesion molecule. Ethanol interacts with an alcohol binding pocket in the L1 extracellular domain (ECD), and dephosphorylation of S1248 in the L1 cytoplasmic domain (CD) renders L1 adhesion insensitive to inhibition by ethanol (L1 insensitive). The mechanism underlying this inside-out signaling is unknown. Here we show that phosphorylation of the human L1-CD at S1152, Y1176, S1181, and S1248 renders L1 sensitive to ethanol by promoting L1 coupling with ankyrin-G and the spectrin-actin cytoskeleton. Knockdown of ankyrin-G or L1 mutations that uncouple L1 from ankyrin reduce L1 sensitivity to ethanol, but not methanol, consistent with a small conformational change in the extracellular alcohol binding pocket. Phosphorylation of Y1176 and ankyrin-G coupling with L1 are higher in NIH/3T3 clonal cell lines in which ethanol inhibits L1 adhesion than in ethanol-resistant NIH/3T3 clonal cell lines. Similarly, phosphorylation of Y1176 is higher in C57BL/6J mice that are sensitive to ethanol teratogenesis than in ethanol resistant C57BL/6N mice. Finally, polymorphisms in genes that encode ankyrin-G and p90rsk, a kinase that phosphorylates S1152, are linked to facial dysmorphology in children with heavy prenatal ethanol exposure. These findings indicate that genes that regulate L1 coupling to ankyrin may influence susceptibility to FASD.-Dou, X., Menkari, C., Mitsuyama, R., Foroud, T., Wetherill, L., Hammond, P., Suttie, M., Chen, X., Chen, S.-Y., Charness, M. E., Collaborative Initiative on Fetal Alcohol Spectrum Disorders. L1 coupling to ankyrin and the spectrin-actin cytoskeleton modulates ethanol inhibition of L1 adhesion and ethanol teratogenesis.
Collapse
Affiliation(s)
- Xiaowei Dou
- Department of Neurology, Veterans Affairs Boston Healthcare System, Harvard Medical School, West Roxbury, Massachusetts, USA
| | - Carrie Menkari
- Department of Neurology, Veterans Affairs Boston Healthcare System, Harvard Medical School, West Roxbury, Massachusetts, USA
| | - Rei Mitsuyama
- Department of Neurology, Veterans Affairs Boston Healthcare System, Harvard Medical School, West Roxbury, Massachusetts, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Leah Wetherill
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Peter Hammond
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
| | - Michael Suttie
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
| | - Xiaopan Chen
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Shao-Yu Chen
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, Kentucky, USA; and
| | - Michael E Charness
- Department of Neurology, Veterans Affairs Boston Healthcare System, Harvard Medical School, West Roxbury, Massachusetts, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
18
|
Wells PG, Bhatia S, Drake DM, Miller-Pinsler L. Fetal oxidative stress mechanisms of neurodevelopmental deficits and exacerbation by ethanol and methamphetamine. ACTA ACUST UNITED AC 2017; 108:108-30. [PMID: 27345013 DOI: 10.1002/bdrc.21134] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/09/2016] [Indexed: 11/06/2022]
Abstract
In utero exposure of mouse progeny to alcohol (ethanol, EtOH) and methamphetamine (METH) causes substantial postnatal neurodevelopmental deficits. One emerging pathogenic mechanism underlying these deficits involves fetal brain production of reactive oxygen species (ROS) that alter signal transduction, and/or oxidatively damage cellular macromolecules like lipids, proteins, and DNA, the latter leading to altered gene expression, likely via non-mutagenic mechanisms. Even physiological levels of fetal ROS production can be pathogenic in biochemically predisposed progeny, and ROS formation can be enhanced by drugs like EtOH and METH, via activation/induction of ROS-producing NADPH oxidases (NOX), drug bioactivation to free radical intermediates by prostaglandin H synthases (PHS), and other mechanisms. Antioxidative enzymes, like catalase in the fetal brain, while low, provide critical protection. Oxidatively damaged DNA is normally rapidly repaired, and fetal deficiencies in several DNA repair proteins, including oxoguanine glycosylase 1 (OGG1) and breast cancer protein 1 (BRCA1), enhance the risk of drug-initiated postnatal neurodevelopmental deficits, and in some cases deficits in untreated progeny, the latter of which may be relevant to conditions like autism spectrum disorders (ASD). Risk is further regulated by fetal nuclear factor erythroid 2-related factor 2 (Nrf2), a ROS-sensing protein that upregulates an array of proteins, including antioxidative enzymes and DNA repair proteins. Imbalances between conceptal pathways for ROS formation, versus those for ROS detoxification and DNA repair, are important determinants of risk. Birth Defects Research (Part C) 108:108-130, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Peter G Wells
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Canada.,Department of Pharmacology & Toxicology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Shama Bhatia
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Danielle M Drake
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Lutfiya Miller-Pinsler
- Department of Pharmacology & Toxicology, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
19
|
Base-Mediated One-Pot Synthesis of Aliphatic Diazirines for Photoaffinity Labeling. Molecules 2017; 22:molecules22081389. [PMID: 28829361 PMCID: PMC6152361 DOI: 10.3390/molecules22081389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 01/12/2023] Open
Abstract
Aliphatic diazirines have been widely used as prominent photophores for photoaffinity labeling owing to their relatively small size which can reduce the steric effect on the natural interaction between ligands and proteins. Based on our continuous efforts to develop efficient methods for the synthesis of aliphatic diazirines, we present here a comprehensive study about base-mediated one-pot synthesis of aliphatic diazirines. It was found that potassium hydroxide (KOH) can also promote the construction of aliphatic diazirine with good efficiency. Importantly, KOH is cheaper, highly available, and easily handled and stored compared with the previously used base, potassium tert-butoxide (t-BuOK). Gram-scale study showed that it owned great advantages in being used for the large-scale production of aliphatic diazirines. This protocol is highly neat and the desired products can be easily isolated and purified. As the first comprehensive study of the base-mediated one-pot synthesis of aliphatic diazirines, this work provided good insight into the preparation and utilization of diazirine-based photoaffinity labeling probes.
Collapse
|
20
|
Woll KA, Dailey WP, Brannigan G, Eckenhoff RG. Shedding Light on Anesthetic Mechanisms: Application of Photoaffinity Ligands. Anesth Analg 2017; 123:1253-1262. [PMID: 27464974 DOI: 10.1213/ane.0000000000001365] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Anesthetic photoaffinity ligands have had an increasing presence within anesthesiology research. These ligands mimic parent general anesthetics and allow investigators to study anesthetic interactions with receptors and enzymes; identify novel targets; and determine distribution within biological systems. To date, nearly all general anesthetics used in medicine have a corresponding photoaffinity ligand represented in the literature. In this review, we examine all aspects of the current methodologies, including ligand design, characterization, and deployment. Finally we offer points of consideration and highlight the future outlook as more photoaffinity ligands emerge within the field.
Collapse
Affiliation(s)
- Kellie A Woll
- From the Departments of *Anesthesiology and Critical Care and †Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; ‡Department of Chemistry, University of Pennsylvania School of Arts and Sciences, Philadelphia, Pennsylvania; and §Department of Physics, Rutgers University, Camden, New Jersey
| | | | | | | |
Collapse
|
21
|
Cui C, Koob GF. Titrating Tipsy Targets: The Neurobiology of Low-Dose Alcohol. Trends Pharmacol Sci 2017; 38:556-568. [PMID: 28372826 DOI: 10.1016/j.tips.2017.03.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/04/2017] [Accepted: 03/06/2017] [Indexed: 11/30/2022]
Abstract
Limited attention has been given to our understanding of how the brain responds to low-dose alcohol (ethanol) and what molecular and cellular targets mediate these effects. Even at concentrations lower than 10mM (0.046 g% blood alcohol concentration, BAC), below the legal driving limit in the USA (BAC 0.08 g%), alcohol impacts brain function and behavior. Understanding what molecular and cellular targets mediate the initial effects of alcohol and subsequent neuroplasticity could provide a better understanding of vulnerability or resilience to developing alcohol use disorders. We review here what is known about the neurobiology of low-dose alcohol, provide insights into potential molecular targets, and discuss future directions and challenges in further defining targets of low-dose alcohol at the molecular, cellular, and circuitry levels.
Collapse
Affiliation(s)
- Changhai Cui
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
22
|
Lovely C, Rampersad M, Fernandes Y, Eberhart J. Gene-environment interactions in development and disease. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2017; 6:10.1002/wdev.247. [PMID: 27626243 PMCID: PMC5191946 DOI: 10.1002/wdev.247] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/08/2016] [Accepted: 07/25/2016] [Indexed: 12/17/2022]
Abstract
Developmental geneticists continue to make substantial jumps in our understanding of the genetic pathways that regulate development. This understanding stems predominantly from analyses of genetically tractable model organisms developing in laboratory environments. This environment is vastly different from that in which human development occurs. As such, most causes of developmental defects in humans are thought to involve multifactorial gene-gene and gene-environment interactions. In this review, we discuss how gene-environment interactions with environmental teratogens may predispose embryos to structural malformations. We elaborate on the growing number of gene-ethanol interactions that might underlie susceptibility to fetal alcohol spectrum disorders. WIREs Dev Biol 2017, 6:e247. doi: 10.1002/wdev.247 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- C Lovely
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Mindy Rampersad
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Yohaan Fernandes
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Johann Eberhart
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
23
|
Patzke C, Acuna C, Giam LR, Wernig M, Südhof TC. Conditional deletion of L1CAM in human neurons impairs both axonal and dendritic arborization and action potential generation. J Exp Med 2016; 213:499-515. [PMID: 27001749 PMCID: PMC4821644 DOI: 10.1084/jem.20150951] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 02/12/2016] [Indexed: 01/02/2023] Open
Abstract
Patzke et al. create human embryonic stem cell–derived neurons that enable the generation of conditional loss-of-function mutations of L1CAM. Deletion of L1CAM impairs axonal elongation, dendritic arborization, and action potential generation. Hundreds of L1CAM gene mutations have been shown to be associated with congenital hydrocephalus, severe intellectual disability, aphasia, and motor symptoms. How such mutations impair neuronal function, however, remains unclear. Here, we generated human embryonic stem (ES) cells carrying a conditional L1CAM loss-of-function mutation and produced precisely matching control and L1CAM-deficient neurons from these ES cells. In analyzing two independent conditionally mutant ES cell clones, we found that deletion of L1CAM dramatically impaired axonal elongation and, to a lesser extent, dendritic arborization. Unexpectedly, we also detected an ∼20–50% and ∼20–30% decrease, respectively, in the levels of ankyrinG and ankyrinB protein, and observed that the size and intensity of ankyrinG staining in the axon initial segment was significantly reduced. Overexpression of wild-type L1CAM, but not of the L1CAM point mutants R1166X and S1224L, rescued the decrease in ankyrin levels. Importantly, we found that the L1CAM mutation selectively decreased activity-dependent Na+-currents, altered neuronal excitability, and caused impairments in action potential (AP) generation. Thus, our results suggest that the clinical presentations of L1CAM mutations in human patients could be accounted for, at least in part, by cell-autonomous changes in the functional development of neurons, such that neurons are unable to develop normal axons and dendrites and to generate normal APs.
Collapse
Affiliation(s)
- Christopher Patzke
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Claudio Acuna
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Louise R Giam
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305 Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305 Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
24
|
Valenzuela CF, Medina AE, Wozniak JR, Klintsova AY. Proceedings of the 2015 Annual Meeting of the Fetal Alcohol Spectrum Disorders Study Group. Alcohol 2016; 50:37-42. [PMID: 26695590 DOI: 10.1016/j.alcohol.2015.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/17/2015] [Accepted: 10/21/2015] [Indexed: 10/22/2022]
Abstract
The 2015 Fetal Alcohol Spectrum Disorders Study Group (FASDSG) meeting was titled "Basic Mechanisms and Translational Implications." Despite decades of basic science and clinical research, our understanding of the mechanisms by which ethanol affects fetal development is still in its infancy. The first keynote presentation focused on the role of heat shock protein pathways in the actions of ethanol in the developing brain. The second keynote presentation addressed the use of magnetoencephalography to characterize brain function in children with FASD. The conference also included talks by representatives from several government agencies, short presentations by junior and senior investigators that showcased the latest in FASD research, and award presentations. An important part of the meeting was the presentation of the 2015 Henry Rosett award to Dr. Michael Charness in honor of his achievements in research on FASD.
Collapse
|
25
|
Chacko AM, Han J, Greineder CF, Zern BJ, Mikitsh JL, Nayak M, Menon D, Johnston IH, Poncz M, Eckmann DM, Davies PF, Muzykantov VR. Collaborative Enhancement of Endothelial Targeting of Nanocarriers by Modulating Platelet-Endothelial Cell Adhesion Molecule-1/CD31 Epitope Engagement. ACS NANO 2015; 9:6785-6793. [PMID: 26153796 PMCID: PMC4761649 DOI: 10.1021/nn505672x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Nanocarriers (NCs) coated with antibodies (Abs) to extracellular epitopes of the transmembrane glycoprotein PECAM (platelet endothelial cell adhesion molecule-1/CD31) enable targeted drug delivery to vascular endothelial cells. Recent studies revealed that paired Abs directed to adjacent, yet distinct epitopes of PECAM stimulate each other's binding to endothelial cells in vitro and in vivo ("collaborative enhancement"). This phenomenon improves targeting of therapeutic fusion proteins, yet its potential role in targeting multivalent NCs has not been addressed. Herein, we studied the effects of Ab-mediated collaborative enhancement on multivalent NC spheres coated with PECAM Abs (Ab/NC, ∼180 nm diameter). We found that PECAM Abs do mutually enhance endothelial cell binding of Ab/NC coated by paired, but not "self" Ab. In vitro, collaborative enhancement of endothelial binding of Ab/NC by paired Abs is modulated by Ab/NC avidity, epitope selection, and flow. Cell fixation, but not blocking of endocytosis, obliterated collaborative enhancement of Ab/NC binding, indicating that the effect is mediated by molecular reorganization of PECAM molecules in the endothelial plasmalemma. The collaborative enhancement of Ab/NC binding was affirmed in vivo. Intravascular injection of paired Abs enhanced targeting of Ab/NC to pulmonary vasculature in mice by an order of magnitude. This stimulatory effect greatly exceeded enhancement of Ab targeting by paired Abs, indicating that '"collaborative enhancement"' effect is even more pronounced for relatively large multivalent carriers versus free Abs, likely due to more profound consequences of positive alteration of epitope accessibility. This phenomenon provides a potential paradigm for optimizing the endothelial-targeted nanocarrier delivery of therapeutic agents.
Collapse
Affiliation(s)
- Ann-Marie Chacko
- Department of Radiology, Division of Nuclear Medicine and Clinical Molecular Imaging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Targeted Therapeutics and Translational Nanomedicine, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jingyan Han
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Colin F. Greineder
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Blaine J. Zern
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - John L. Mikitsh
- Department of Radiology, Division of Nuclear Medicine and Clinical Molecular Imaging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Madhura Nayak
- Department of Radiology, Division of Nuclear Medicine and Clinical Molecular Imaging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Divya Menon
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ian H. Johnston
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Mortimer Poncz
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - David M. Eckmann
- Department of Anesthesiology & Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Peter F. Davies
- Department of Pathology and Institute for Medicine and Engineering, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R. Muzykantov
- Center for Targeted Therapeutics and Translational Nanomedicine, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
26
|
Dou X, Charness ME. Effect of lipid raft disruption on ethanol inhibition of l1 adhesion. Alcohol Clin Exp Res 2015; 38:2707-11. [PMID: 25421507 PMCID: PMC4278581 DOI: 10.1111/acer.12556] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/24/2014] [Indexed: 12/21/2022]
Abstract
Background Alcohol causes fetal alcohol spectrum disorders in part by disrupting the function of the neural cell adhesion molecule L1. Alcohol inhibits L1-mediated cell–cell adhesion in diverse cell types and inhibits L1-mediated neurite outgrowth in cerebellar granule neurons (CGNs). A recent report indicates that ethanol (EtOH) induces the translocation of L1 into CGN lipid rafts and that disruption of lipid rafts prevents EtOH inhibition of L1-mediated neurite outgrowth. The same butanol–pentanol cutoff was noted for alcohol-induced translocation of L1 into lipid rafts that was reported previously for alcohol inhibition of L1 adhesion, suggesting that EtOH might inhibit L1 adhesion by shifting L1 into lipid rafts. Methods The NIH/3T3 cell line, 2A2-L1s, is a well-characterized EtOH-sensitive clonal cell line that stably expresses human L1. Cells were treated with 25 mM EtOH, 5 μM filipin, or both. Lipid rafts were enriched in membrane fractions by preparation of detergent-resistant membrane (DRMs) fractions. Caveolin-1 was used as a marker of lipid rafts, and L1 and Src were quantified by Western blotting in lipid-raft-enriched membrane fractions and by immunohistochemistry. Results EtOH (25 mM) increased the percentage of L1, but not Src, in 2A2-L1s membrane fractions enriched in lipid rafts. Filipin, an agent known to disrupt lipid rafts, decreased the percentage of caveolin and L1 in DRMs from 2A2-L1s cells. Filipin also blocked EtOH-induced translocation of L1 into lipid rafts from 2A2-L1s cells but did not significantly affect L1 adhesion or EtOH inhibition of L1 adhesion. Conclusions These findings indicate that EtOH does not inhibit L1 adhesion in NIH/3T3 cells by inducing the translocation of L1 into lipid rafts.
Collapse
Affiliation(s)
- Xiaowei Dou
- VA Boston Healthcare System, Harvard Medical School, West Roxbury, Massachusetts
| | | |
Collapse
|
27
|
Karunamuni GH, Ma P, Gu S, Rollins AM, Jenkins MW, Watanabe M. Connecting teratogen-induced congenital heart defects to neural crest cells and their effect on cardiac function. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2014; 102:227-50. [PMID: 25220155 PMCID: PMC4238913 DOI: 10.1002/bdrc.21082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 08/26/2014] [Indexed: 12/26/2022]
Abstract
Neural crest cells play many key roles in embryonic development, as demonstrated by the abnormalities that result from their specific absence or dysfunction. Unfortunately, these key cells are particularly sensitive to abnormalities in various intrinsic and extrinsic factors, such as genetic deletions or ethanol-exposure that lead to morbidity and mortality for organisms. This review discusses the role identified for a segment of neural crest in regulating the morphogenesis of the heart and associated great vessels. The paradox is that their derivatives constitute a small proportion of cells to the cardiovascular system. Findings supporting that these cells impact early cardiac function raises the interesting possibility that they indirectly control cardiovascular development at least partially through regulating function. Making connections between insults to the neural crest, cardiac function, and morphogenesis is more approachable with technological advances. Expanding our understanding of early functional consequences could be useful in improving diagnosis and testing therapies.
Collapse
Affiliation(s)
- Ganga H. Karunamuni
- Department of Pediatrics, Case Western Reserve University School of Medicine, Case Medical Center Division of Pediatric Cardiology, Rainbow Babies and Children’s Hospital, Cleveland OH 44106
| | - Pei Ma
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland OH 44106
| | - Shi Gu
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland OH 44106
| | - Andrew M. Rollins
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland OH 44106
| | - Michael W. Jenkins
- Department of Pediatrics, Case Western Reserve University School of Medicine, Case Medical Center Division of Pediatric Cardiology, Rainbow Babies and Children’s Hospital, Cleveland OH 44106
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland OH 44106
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University School of Medicine, Case Medical Center Division of Pediatric Cardiology, Rainbow Babies and Children’s Hospital, Cleveland OH 44106
| |
Collapse
|
28
|
Mei H, Campbell JM, Paddock CM, Lertkiatmongkol P, Mosesson MW, Albrecht R, Newman PJ. Regulation of endothelial cell barrier function by antibody-driven affinity modulation of platelet endothelial cell adhesion molecule-1 (PECAM-1). J Biol Chem 2014; 289:20836-44. [PMID: 24936065 PMCID: PMC4110291 DOI: 10.1074/jbc.m114.557454] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 06/10/2014] [Indexed: 12/31/2022] Open
Abstract
PECAM-1 is a 130-kDa member of the immunoglobulin (Ig) superfamily that is expressed on the surface of platelets and leukocytes, and at the intracellular junctions of confluent endothelial cell monolayers. Previous studies have shown that PECAM-1/PECAM-1 homophilic interactions play a key role in leukocyte transendothelial migration, in allowing PECAM-1 to serve as a mechanosensory complex in endothelial cells, in its ability to confer cytoprotection to proapoptotic stimuli, and in maintaining endothelial cell junctional integrity. To examine the adhesive properties of full-length PECAM-1 in a native lipid environment, we purified it from platelets and assembled it into phospholipid nanodiscs. PECAM-1-containing nanodiscs retained not only their ability to bind homophilically to PECAM-1-expressing cells, but exhibited regulatable adhesive interactions that could be modulated by ligands that bind membrane- proximal Ig Domain 6. This property was exploited to enhance the rate of barrier restoration in endothelial cell monolayers subjected to inflammatory challenge. The finding that the adhesive properties of PECAM-1 are regulatable suggests novel approaches for controlling endothelial cell migration and barrier function in a variety of vascular permeability disorders.
Collapse
Affiliation(s)
- Heng Mei
- From the Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin 53226
| | | | - Cathy M. Paddock
- From the Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin 53226
| | - Panida Lertkiatmongkol
- From the Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin 53226
- the Department of Pharmacology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Michael W. Mosesson
- From the Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin 53226
| | - Ralph Albrecht
- the Departments of Animal Sciences, Pediatrics, and Pharmaceutical Sciences, University of Wisconsin, Madison, Wisconsin 54701, and
| | - Peter J. Newman
- From the Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Wisconsin 53226
- the Department of Pharmacology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
29
|
Das J, Xu S, Pany S, Guillory A, Shah V, Roman GW. The pre-synaptic Munc13-1 binds alcohol and modulates alcohol self-administration in Drosophila. J Neurochem 2013; 126:715-26. [PMID: 23692447 DOI: 10.1111/jnc.12315] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/09/2013] [Accepted: 05/17/2013] [Indexed: 11/30/2022]
Abstract
Munc13-1 is a pre-synaptic active-zone protein essential for neurotransmitter release and involved in pre-synaptic plasticity in brain. Ethanol, butanol, and octanol quenched the intrinsic fluorescence of the C1 domain of Munc13-1 with EC₅₀ s of 52 mM, 26 mM, and 0.7 mM, respectively. Photoactive azialcohols photolabeled Munc13-1 C1 exclusively at Glu-582, which was identified by mass spectrometry. Mutation of Glu-582 to alanine, leucine, and histidine reduced the alcohol binding two- to five-fold. Circular dichroism studies suggested that binding of alcohol increased the stability of the wild-type Munc13-1 compared with the mutants. If Munc13-1 plays some role in the neural effects of alcohol in vivo, changes in the activity of this protein should produce differences in the behavioral responses to ethanol. We tested this prediction with a loss-of-function mutation in the conserved Dunc-13 in Drosophila melanogaster. The Dunc-13(P84200) /+ heterozygotes have 50% wild-type levels of Dunc-13 mRNA and display a very robust increase in ethanol self-administration. This phenotype is reversed by the expression of the rat Munc13-1 protein within the Drosophila nervous system. The present studies indicate that Munc13-1 C1 has binding site(s) for alcohols and Munc13-1 activity is sufficient to restore normal self-administration to Drosophila mutants deficient in Dunc-13 activity. The pre-synaptic Mun13-1 protein is a critical regulator of synaptic vesicle fusion and may be involved in processes that lead to ethanol abuse and addiction. We studied its interaction with alcohol and identified Glu-582 as a critical residue for ethanol binding. Munc13-1 can functionally complement the Dunc13 haploinsufficient ethanol self-administration phenotype in Drosophila melanogaster, indicating that this protein participates in alcohol-induced behavioral plasticity.
Collapse
Affiliation(s)
- Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas 77204, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Homophilic interaction of the L1 family of cell adhesion molecules plays a pivotal role in regulating neurite outgrowth and neural cell networking in vivo. Functional defects in L1 family members are associated with neurological disorders such as X-linked mental retardation, multiple sclerosis, low-IQ syndrome, developmental delay, and schizophrenia. Various human tumors with poor prognosis also implicate the role of L1, a representative member of the L1 family of cell adhesion molecules, and ectopic expression of L1 in fibroblastic cells induces metastasis-associated gene expression. Previous studies on L1 homologs indicated that four N-terminal immunoglobulin-like domains form a horseshoe-like structure that mediates homophilic interactions. Various models including the zipper, domain-swap, and symmetry-related models are proposed to be involved in structural mechanism of homophilic interaction of the L1 family members. Recently, cryo-electron tomography of L1 and crystal structure studies of neurofascin, an L1 family protein, have been performed. This review focuses on recent discoveries of different models and describes the possible structural mechanisms of homophilic interactions of L1 family members. Understanding structural mechanisms of homophilic interactions in various cell adhesion proteins should aid the development of therapeutic strategies for L1 family cell adhesion molecule-associated diseases.
Collapse
Affiliation(s)
- Chun Hua Wei
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, Korea
| | | |
Collapse
|
31
|
|
32
|
Mitogen-activated protein kinase modulates ethanol inhibition of cell adhesion mediated by the L1 neural cell adhesion molecule. Proc Natl Acad Sci U S A 2013; 110:5683-8. [PMID: 23431142 DOI: 10.1073/pnas.1221386110] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
There is a genetic contribution to fetal alcohol spectrum disorders (FASD), but the identification of candidate genes has been elusive. Ethanol may cause FASD in part by decreasing the adhesion of the developmentally critical L1 cell adhesion molecule through interactions with an alcohol binding pocket on the extracellular domain. Pharmacologic inhibition or genetic knockdown of ERK2 did not alter L1 adhesion, but markedly decreased ethanol inhibition of L1 adhesion in NIH/3T3 cells and NG108-15 cells. Likewise, leucine replacement of S1248, an ERK2 substrate on the L1 cytoplasmic domain, did not decrease L1 adhesion, but abolished ethanol inhibition of L1 adhesion. Stable transfection of NIH/3T3 cells with human L1 resulted in clonal cell lines in which L1 adhesion was consistently sensitive or insensitive to ethanol for more than a decade. ERK2 activity and S1248 phosphorylation were greater in ethanol-sensitive NIH/3T3 clonal cell lines than in their ethanol-insensitive counterparts. Ethanol-insensitive cells became ethanol sensitive after increasing ERK2 activity by transfection with a constitutively active MAP kinase kinase 1. Finally, embryos from two substrains of C57BL mice that differ in susceptibility to ethanol teratogenesis showed corresponding differences in MAPK activity. Our data suggest that ERK2 phosphorylation of S1248 modulates ethanol inhibition of L1 adhesion by inside-out signaling and that differential regulation of ERK2 signaling might contribute to genetic susceptibility to FASD. Moreover, identification of a specific locus that regulates ethanol sensitivity, but not L1 function, might facilitate the rational design of drugs that block ethanol neurotoxicity.
Collapse
|
33
|
Tagliavacca L, Colombo F, Racchetti G, Meldolesi J. L1CAM and its cell-surface mutants: new mechanisms and effects relevant to the physiology and pathology of neural cells. J Neurochem 2012; 124:397-409. [PMID: 22973895 PMCID: PMC3557714 DOI: 10.1111/jnc.12015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 09/05/2012] [Accepted: 09/08/2012] [Indexed: 11/27/2022]
Abstract
The L1 syndrome, a genetic disease that affects 1/30 000 newborn males, is sustained by numerous missense mutations of L1 cell adhesion molecule (L1CAM), an adhesion surface protein active also in transmembrane signaling, essential for the development and function of neurons. To investigate the cell biology of L1CAM, we employed a high RE1-silencing transcription (factor) clone of the pheochromocytoma PC12 line, defective in L1CAM expression and neurite outgrowth. The clone was transfected with wild-type L1CAM and four missense, disease-inducing point mutants encoding proteins distributed to the cell surface. The mutant-expressing cells, defective in adhesion to extracellular matrix proteins and in migration, exhibited unchanged proliferation. The nerve growth factor (NGF)-induced neurite outgrowth was re-established in defective clone cells transfected with the wild-type and the H210Q and I219T L1CAMs mutants, but not in the others. The stimulated outgrowth was confirmed in a second defective PC12 clone over-expressing the NGF receptor TrkA, treated with NGF and/or a recombinant L1CAM chimera. These results revealed a new function of L1CAM, a positive, robust and dose-dependent modulation of the TrkA receptor activated spontaneously or by NGF. The variable effects observed with the different L1CAM mutants suggest that this function contributes to the marked heterogeneity of symptoms and severity observed in the patients affected by the L1 syndrome.
Collapse
Affiliation(s)
- Luigina Tagliavacca
- Department of Neuroscience, Vita-Salute San Raffaele University and San Raffaele Institute, Milano, Italy
| | | | | | | |
Collapse
|
34
|
Kapfhamer D, Taylor S, Zou ME, Lim JP, Kharazia V, Heberlein U. Taok2 controls behavioral response to ethanol in mice. GENES BRAIN AND BEHAVIOR 2012; 12:87-97. [PMID: 22883308 DOI: 10.1111/j.1601-183x.2012.00834.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/30/2012] [Accepted: 08/02/2012] [Indexed: 01/27/2023]
Abstract
Despite recent advances in the understanding of ethanol's biological action, many of the molecular targets of ethanol and mechanisms behind ethanol's effect on behavior remain poorly understood. In an effort to identify novel genes, the products of which regulate behavioral responses to ethanol, we recently identified a mutation in the dtao gene that confers resistance to the locomotor stimulating effect of ethanol in Drosophila. dtao encodes a member of the Ste20 family of serine/threonine kinases implicated in MAP kinase signaling pathways. In this study, we report that conditional ablation of the mouse dtao homolog, Taok2, constitutively and specifically in the nervous system, results in strain-specific and overlapping alterations in ethanol-dependent behaviors. These data suggest a functional conservation of dtao and Taok2 in mediating ethanol's biological action and identify Taok2 as a putative candidate gene for ethanol use disorders in humans.
Collapse
Affiliation(s)
- D Kapfhamer
- The Ernest Gallo Clinic and Research Center, University of California at San Francisco, Emeryville, CA 94608, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Petit-Paitel A, Ménard B, Guyon A, Béringue V, Nahon JL, Zsürger N, Chabry J. Prion protein is a key determinant of alcohol sensitivity through the modulation of N-methyl-D-aspartate receptor (NMDAR) activity. PLoS One 2012; 7:e34691. [PMID: 22536327 PMCID: PMC3335038 DOI: 10.1371/journal.pone.0034691] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 03/06/2012] [Indexed: 11/21/2022] Open
Abstract
The prion protein (PrP) is absolutely required for the development of prion diseases; nevertheless, its physiological functions in the central nervous system remain elusive. Using a combination of behavioral, electrophysiological and biochemical approaches in transgenic mouse models, we provide strong evidence for a crucial role of PrP in alcohol sensitivity. Indeed, PrP knock out (PrP−/−) mice presented a greater sensitivity to the sedative effects of EtOH compared to wild-type (wt) control mice. Conversely, compared to wt mice, those over-expressing mouse, human or hamster PrP genes presented a relative insensitivity to ethanol-induced sedation. An acute tolerance (i.e. reversion) to ethanol inhibition of N-methyl-D-aspartate (NMDA) receptor-mediated excitatory post-synaptic potentials in hippocampal slices developed slower in PrP−/− mice than in wt mice. We show that PrP is required to induce acute tolerance to ethanol by activating a Src-protein tyrosine kinase-dependent intracellular signaling pathway. In an attempt to decipher the molecular mechanisms underlying PrP-dependent ethanol effect, we looked for changes in lipid raft features in hippocampus of ethanol-treated wt mice compared to PrP−/− mice. Ethanol induced rapid and transient changes of buoyancy of lipid raft-associated proteins in hippocampus of wt but not PrP−/− mice suggesting a possible mechanistic link for PrP-dependent signal transduction. Together, our results reveal a hitherto unknown physiological role of PrP on the regulation of NMDAR activity and highlight its crucial role in synaptic functions.
Collapse
Affiliation(s)
- Agnès Petit-Paitel
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Baptiste Ménard
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Alice Guyon
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Vincent Béringue
- Institut National de la Recherche Agronomique, UR892, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Jean-Louis Nahon
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Nicole Zsürger
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Joëlle Chabry
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- * E-mail:
| |
Collapse
|
36
|
Désy O, Carignan D, de Campos-Lima PO. Short-term immunological effects of non-ethanolic short-chain alcohols. Toxicol Lett 2012; 210:44-52. [PMID: 22266471 DOI: 10.1016/j.toxlet.2012.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 01/05/2012] [Accepted: 01/05/2012] [Indexed: 10/14/2022]
Abstract
Short-chain alcohols are embedded into several aspects of modern life. The societal costs emanating from the long history of use and abuse of the prototypical example of these molecules, ethanol, have stimulated considerable interest in its general toxicology. A much more modest picture exists for other short-chain alcohols, notably as regards their immunotoxicity. A large segment of the general population is potentially exposed to two of these alcohols, methanol and isopropanol. Their ubiquitous nature and their eventual use as ethanol surrogates are predictably associated to accidental or deliberate poisoning. This review addresses the immunological consequences of acute exposure to methanol and isopropanol. It first examines the general mechanisms of short-chain alcohol-induced biological dysregulation and then provides a tentative model to explain the molecular events that underlie the immunological dysfunction produced by methanol and isopropanol. The time-related context of serum alcohol concentrations in acute poisoning, as well as the clinical implications of their short-term immunotoxicity, is also discussed.
Collapse
Affiliation(s)
- Olivier Désy
- Laval University Cancer Research Center, Quebec City, Quebec, Canada
| | | | | |
Collapse
|
37
|
Alfonso-Loeches S, Guerri C. Molecular and behavioral aspects of the actions of alcohol on the adult and developing brain. Crit Rev Clin Lab Sci 2011; 48:19-47. [PMID: 21657944 DOI: 10.3109/10408363.2011.580567] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The brain is one of the major target organs of alcohol actions. Alcohol abuse can lead to alterations in brain structure and functions and, in some cases, to neurodegeneration. Cognitive deficits and alcohol dependence are highly damaging consequences of alcohol abuse. Clinical and experimental studies have demonstrated that the developing brain is particularly vulnerable to alcohol, and that drinking during gestation can lead to a range of physical, learning and behavioral defects (fetal alcohol spectrum disorders), with the most dramatic presentation corresponding to fetal alcohol syndrome. Recent findings also indicate that adolescence is a stage of brain maturation and that heavy drinking at this stage can have a negative impact on brain structure and functions causing important short- and long-term cognitive and behavioral consequences. The effects of alcohol on the brain are not uniform; some brain areas or cell populations are more vulnerable than others. The prefrontal cortex, the hippocampus, the cerebellum, the white matter and glial cells are particularly susceptible to the effects of ethanol. The molecular actions of alcohol on the brain are complex and involve numerous mechanisms and signaling pathways. Some of the mechanisms involved are common for the adult brain and for the developing brain, while others depend on the developmental stage. During brain ontogeny, alcohol causes irreversible alterations to the brain structure. It also impairs several molecular, neurochemical and cellular events taking place during normal brain development, including alterations in both gene expression regulation and the molecules involved in cell-cell interactions, interference with the mitogenic and growth factor response, enhancement of free radical formation and derangements of glial cell functions. However, in both adult and adolescent brains, alcohol damages specific brain areas through mechanisms involving excitotoxicity, free radical formation and neuroinflammatory damage resulting from activation of the innate immune system mediated by TLR4 receptors. Alcohol also acts on specific membrane proteins, such as neurotransmitter receptors (e.g. NMDA, GABA-A), ion channels (e.g. L-type Ca²⁺ channels, GIRKs), and signaling pathways (e.g. PKA and PKC signaling). These effects might underlie the wide variety of behavioral effects induced by ethanol drinking. The neuroadaptive changes affecting neurotransmission systems which are more sensitive to the acute effects of alcohol occur after long-term alcohol consumption. Alcohol-induced maladaptations in the dopaminergic mesolimbic system, abnormal plastic changes in the reward-related brain areas and genetic and epigenetic factors may all contribute to alcohol reinforcement and alcohol addiction. This manuscript reviews the mechanisms by which ethanol impacts the adult and the developing brain, and causes both neural impairments and cognitive and behavioral dysfunctions. The identification and the understanding of the cellular and molecular mechanisms involved in ethanol toxicity might contribute to the development of treatments and/or therapeutic agents that could reduce or eliminate the deleterious effects of alcohol on the brain.
Collapse
|
38
|
Fitzgerald DM, Charness ME, Leite-Morris KA, Chen S. Effects of ethanol and NAP on cerebellar expression of the neural cell adhesion molecule L1. PLoS One 2011; 6:e24364. [PMID: 21931691 PMCID: PMC3169602 DOI: 10.1371/journal.pone.0024364] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 08/08/2011] [Indexed: 01/16/2023] Open
Abstract
The neural cell adhesion molecule L1 is critical for brain development and plays a role in learning and memory in the adult. Ethanol inhibits L1-mediated cell adhesion and neurite outgrowth in cerebellar granule neurons (CGNs), and these actions might underlie the cerebellar dysmorphology of fetal alcohol spectrum disorders. The peptide NAP potently blocks ethanol inhibition of L1 adhesion and prevents ethanol teratogenesis. We used quantitative RT-PCR and Western blotting of extracts of cerebellar slices, CGNs, and astrocytes from postnatal day 7 (PD7) rats to investigate whether ethanol and NAP act in part by regulating the expression of L1. Treatment of cerebellar slices with 20 mM ethanol, 10−12 M NAP, or both for 4 hours, 24 hours, and 10 days did not significantly affect L1 mRNA and protein levels. Similar treatment for 4 or 24 hours did not regulate L1 expression in primary cultures of CGNs and astrocytes, the predominant cerebellar cell types. Because ethanol also damages the adult cerebellum, we studied the effects of chronic ethanol exposure in adult rats. One year of binge drinking did not alter L1 gene and protein expression in extracts from whole cerebellum. Thus, ethanol does not alter L1 expression in the developing or adult cerebellum; more likely, ethanol disrupts L1 function by modifying its conformation and signaling. Likewise, NAP antagonizes the actions of ethanol without altering L1 expression.
Collapse
Affiliation(s)
- Devon M. Fitzgerald
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts, United States of America
| | - Michael E. Charness
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts, United States of America
- Department of Neurology, Harvard Medical School, West Roxbury, Massachusetts, United States of America
- Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Kimberly A. Leite-Morris
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts, United States of America
- Departments of Psychiatry, Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Suzhen Chen
- Department of Neurology, Harvard Medical School, West Roxbury, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
39
|
|
40
|
Sepulveda B, Carcea I, Zhao B, Salton SR, Benson DL. L1 cell adhesion molecule promotes resistance to alcohol-induced silencing of growth cone responses to guidance cues. Neuroscience 2011; 180:30-40. [PMID: 21335065 PMCID: PMC3070798 DOI: 10.1016/j.neuroscience.2011.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 01/20/2011] [Accepted: 02/08/2011] [Indexed: 01/05/2023]
Abstract
Alcohol exposure in utero is a common cause of mental retardation, but the targets and mechanisms of action are poorly understood. Several lines of data point toward alterations in cortical connectivity, suggesting that axon guidance may be vulnerable to alcohol exposure. To test this, we asked whether ethanol directly affects cortical axonal growth cone responses to guidance cues. We find that even low concentrations of ethanol (12.5 mM; 57.2 mg/dl) commonly observed in social drinking prevent growth cone responses to three mechanistically independent guidance cues, Semaphorin3A, Lysophosphatidic Acid, and Netrin-1. However, this effect is highly dependent on substrate; axonal growth cones extending on an L1 cell adhesion molecule (L1CAM) substrate retain responsiveness to cues following exposure to ethanol, while those growing on poly-L-lysine or N-cadherin do not. The effects of ethanol on axon extension are, by contrast, quite modest. Quantitative assessments of the effects of ethanol on the surface distribution of L1CAM in growth cones suggest that L1CAM homophilic interactions may be particularly relevant for retaining growth cone responsiveness following ethanol exposure. Together, our findings indicate that ethanol can directly and generally alter growth cone responses to guidance cues, that a substrate of L1CAM effectively antagonizes this effect, and that cortical axonal growth cone vulnerability to ethanol may be predicted in part based on the environment through which they are extending.
Collapse
Affiliation(s)
- Bryan Sepulveda
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Ioana Carcea
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Becky Zhao
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Stephen R.J. Salton
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
- Brookdale Department of Geriatrics and Palliative Medicine, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Deanna L. Benson
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| |
Collapse
|
41
|
Das J. Aliphatic diazirines as photoaffinity probes for proteins: recent developments. Chem Rev 2011; 111:4405-17. [PMID: 21466226 DOI: 10.1021/cr1002722] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas 77204, USA.
| |
Collapse
|
42
|
Dou X, Menkari CE, Shanmugasundararaj S, Miller KW, Charness ME. Two alcohol binding residues interact across a domain interface of the L1 neural cell adhesion molecule and regulate cell adhesion. J Biol Chem 2011; 286:16131-9. [PMID: 21367865 DOI: 10.1074/jbc.m110.209254] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ethanol may cause fetal alcohol spectrum disorders (FASD) in part by inhibiting cell adhesion mediated by the L1 neural cell adhesion molecule. Azialcohols photolabel Glu-33 and Tyr-418, two residues that are predicted by homology modeling to lie within 2.8 Å of each other at the interface between the Ig1 and Ig4 domains of L1 (Arevalo, E., Shanmugasundararaj, S., Wilkemeyer, M. F., Dou, X., Chen, S., Charness, M. E., and Miller, K. W. (2008) Proc. Natl. Acad. Sci. U.S.A. 105, 371-375). Using transient transfection of NIH/3T3 cells with wild type (WT-L1) and mutated L1, we found that cysteine substitution of both residues (E33C/Y418C-L1) significantly increased L1 adhesion above levels observed for WT-L1 or the single cysteine substitutions E33C-L1 or Y418C-L1. The reducing agent β-mercaptoethanol (βME) reversibly decreased the adhesion of E33C/Y418C-L1, but had no effect on WT-L1, E33C-L1, or Y418C-L1. Thus, disulfide bond formation occurs between Cys-33 and Cys-418, confirming both the close proximity of these residues and the importance of Ig1-Ig4 interactions in L1 adhesion. Maximal ethanol inhibition of cell adhesion was significantly lower in cells expressing E33C/Y418C-L1 than in those expressing WT-L1, E33C-L1, or Y418C-L1. Moreover, the effects of βME and ethanol on E33C/Y418C-L1 adhesion were non-additive. The cutoff for alcohol inhibition of WT-L1 adhesion was between 1-butanol and 1-pentanol. Increasing the size of the alcohol binding pocket by mutating Glu-33 to Ala-33, increased the alcohol cutoff from 1-butanol to 1-decanol. These findings support the hypothesis that alcohol binding within a pocket bordered by Glu-33 and Tyr-418 inhibits L1 adhesion by disrupting the Ig1-Ig4 interaction.
Collapse
Affiliation(s)
- Xiaowei Dou
- Veterans Affairs Boston Healthcare System, Department of Neurology, Harvard Medical School, West Roxbury, Massachusetts 02132, USA
| | | | | | | | | |
Collapse
|
43
|
Idrus NM, Thomas JD. Fetal alcohol spectrum disorders: experimental treatments and strategies for intervention. ALCOHOL RESEARCH & HEALTH : THE JOURNAL OF THE NATIONAL INSTITUTE ON ALCOHOL ABUSE AND ALCOHOLISM 2011; 34:76-85. [PMID: 23580044 PMCID: PMC3831616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/30/2022]
Abstract
Despite the known damaging effects of prenatal alcohol exposure, women continue to drink during pregnancy, creating a need for effective interventions and treatments for fetal alcohol spectrum disorders (FASD). Experimental models can be useful in identifying potential treatments, and this article describes the spectrum of experimental therapeutics that currently are being investigated, including pharmacological, nutritional, and environmental/behavioral interventions. Some treatments target the underlying mechanisms that contribute to alcohol-induced damage, protecting against alcohol's teratogenic effects, whereas other treatments may enhance central nervous system plasticity either during alcohol exposure or long after alcohol exposure has ceased. The insights gained to date from experimental models offer several candidates for attenuating the deficits associated with FASD.
Collapse
|
44
|
Goodlett CR. Fetal alcohol spectrum disorders: new perspectives on diagnosis and intervention. Alcohol 2010; 44:579-82. [PMID: 21112470 DOI: 10.1016/j.alcohol.2010.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 10/19/2010] [Indexed: 11/26/2022]
|
45
|
Désy O, Carignan D, Caruso M, de Campos-Lima PO. Methanol induces a discrete transcriptional dysregulation that leads to cytokine overproduction in activated lymphocytes. Toxicol Sci 2010; 117:303-13. [PMID: 20616203 DOI: 10.1093/toxsci/kfq212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Methanol is an important cause of acute alcohol intoxication; it is ubiquitously present at home and in the workplace. Although the existing literature provides a reasonable insight into the immunological impact of ethanol and to a much lesser extent of isopropanol, much less data are available on methanol. We hypothesized on structural grounds that methanol would share the immunosuppressive properties of the two other short-chain alcohols. We report here that methanol increases the proliferative capacity of human T lymphocytes and synergizes with the activating stimuli to augment cytokine production. The cytokine upregulation was observed in vitro at methanol concentrations as low as 0.08% (25mM) as measured by interleukin-2, interferon-γ, and tumor necrosis factor-α release in T cells. Methanol did not affect the antigen receptor-mediated early signaling but promoted a selective and differential activation of the nuclear factor of activated T cells family of transcription factors. These results were further substantiated in a mouse model of acute methanol intoxication in which there was an augmented release of proinflammatory cytokines in the serum in response to the staphylococcal enterotoxin B. Our results suggest that methanol has a discrete immunological footprint of broad significance given the exposure of the general population to this multipurpose solvent.
Collapse
Affiliation(s)
- Olivier Désy
- Laval University Cancer Research Center, Quebec City, Quebec, Canada G1R 2J6
| | | | | | | |
Collapse
|
46
|
Schäfer MKE, Altevogt P. L1CAM malfunction in the nervous system and human carcinomas. Cell Mol Life Sci 2010; 67:2425-37. [PMID: 20237819 PMCID: PMC11115577 DOI: 10.1007/s00018-010-0339-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 01/30/2010] [Accepted: 02/11/2010] [Indexed: 12/14/2022]
Abstract
Research over the last 25 years on the cell adhesion molecule L1 has revealed its pivotal role in nervous system function. Mutations of the human L1CAM gene have been shown to cause neurodevelopmental disorders such as X-linked hydrocephalus, spastic paraplegia and mental retardation. Impaired L1 function has been also implicated in the aetiology of fetal alcohol spectrum disorders, defective enteric nervous system development and malformations of the renal system. Importantly, aberrant expression of L1 has emerged as a critical factor in the development of human carcinomas, where it enhances cell proliferation, motility and chemoresistance. This discovery promoted collaborative work between tumour biologists and neurobiologists, which has led to a substantial expansion of the basic knowledge about L1 function and regulation. Here we provide an overview of the pathological conditions caused by L1 malfunction. We further discuss how the available data on gene regulation, molecular interactions and posttranslational processing of L1 may contribute to a better understanding of associated neurological and cancerous diseases.
Collapse
Affiliation(s)
- Michael K E Schäfer
- Center for Neurosciences, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany.
| | | |
Collapse
|
47
|
Sasabe T, Ishiura S. Alcoholism and alternative splicing of candidate genes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2010; 7:1448-66. [PMID: 20617039 PMCID: PMC2872348 DOI: 10.3390/ijerph7041448] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 03/21/2010] [Accepted: 03/23/2010] [Indexed: 11/24/2022]
Abstract
Gene expression studies have shown that expression patterns of several genes have changed during the development of alcoholism. Gene expression is regulated not only at the level of transcription but also through alternative splicing of pre-mRNA. In this review, we discuss some of the evidence suggesting that alternative splicing of candidate genes such as DRD2 (encoding dopamine D2 receptor) may form the basis of the mechanisms underlying the pathophysiology of alcoholism. These reports suggest that aberrant expression of splice variants affects alcohol sensitivities, and alcohol consumption also regulates alternative splicing. Thus, investigations of alternative splicing are essential for understanding the molecular events underlying the development of alcoholism.
Collapse
Affiliation(s)
- Toshikazu Sasabe
- Department of Life Sciences, Graduate School of Arts and Sciences, the University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
| | | |
Collapse
|
48
|
Isayama RN, Leite PEC, Lima JPM, Uziel D, Yamasaki EN. Impact of ethanol on the developing GABAergic system. Anat Rec (Hoboken) 2010; 292:1922-39. [PMID: 19943346 DOI: 10.1002/ar.20966] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alcohol intake during pregnancy has a tremendous impact on the developing brain. Embryonic and early postnatal alcohol exposures have been investigated experimentally to elucidate the fetal alcohol spectrum disorders' (FASD) milieu, and new data have emerged to support a devastating effect on the GABAergic system in the adult and developing nervous system. GABA is a predominantly inhibitory neurotransmitter that during development excites neurons and orchestrates several developmental processes such as proliferation, migration, differentiation, and synaptogenesis. This review summarizes and brings new data on neurodevelopmental aspects of the GABAergic system with FASD in experimental telencephalic models.
Collapse
Affiliation(s)
- Ricardo Noboro Isayama
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
49
|
Fetal alcohol spectrum disorders: from research to policy. ALCOHOL RESEARCH & HEALTH : THE JOURNAL OF THE NATIONAL INSTITUTE ON ALCOHOL ABUSE AND ALCOHOLISM 2010; 33:118-26. [PMID: 23579942 PMCID: PMC3887502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Forty years ago, alcohol was not commonly recognized as a teratogen, an agent that can disrupt the development of a fetus. Today, we understand that prenatal alcohol exposure induces a variety of adverse effects on physical, neurological, and behavioral development. Research supported by the National Institute on Alcohol Abuse and Alcoholism (NIAAA) has contributed to the identification of the range and prevalence of fetal alcohol spectrum disorders (FASD), as well as methods for prevention and treatment of FASD. The worldwide prevalence and high personal and societal costs of FASD speak to the importance of this research. This article briefly examines some of the ways that NIAAA has contributed to our understanding of FASD, the challenges that we still face, and how this research is translated into changes in public policy.
Collapse
|
50
|
Hyun S, Han A, Yu J. Photocrosslinking of RNA and photoMet-containing amphiphilic alpha-helical peptides. Chembiochem 2009; 10:987-9. [PMID: 19308928 DOI: 10.1002/cbic.200900100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Discovering RNA-protein interactions: A library of photoMet-containing peptides was synthesized by using an Arg- and Leu-rich alpha-helical amphiphilic peptide. Irradiation of mixtures of these peptides and Rev-responsive element (RRE) hairpin RNA promoted formation of covalent adducts. Analysis of one adduct showed that U26 in the bulged stem is responsible for covalent bond formation with the carbene intermediate. This strategy can provide important structural information about RNA-peptide interactions.
Collapse
Affiliation(s)
- Soonsil Hyun
- Department of Chemistry and Education, Seoul National University, Korea
| | | | | |
Collapse
|